Home>>Signaling Pathways>> Ubiquitination/ Proteasome>> Autophagy>>DMOG

DMOG Sale

(Synonyms: 二甲基乙二酰氨基乙酸,Dimethyloxallyl Glycine) 目录号 : GC16973

An HIF-PH inhibitor

DMOG Chemical Structure

Cas No.:89464-63-1

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥620.00
现货
50mg
¥462.00
现货
100mg
¥693.00
现货
200mg
¥1,292.00
现货

电话:400-920-5774 Email: sales@glpbio.cn

Customer Reviews

Based on customer reviews.

Sample solution is provided at 25 µL, 10mM.

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

B1 cells

Preparation Method

B1 cells were FACS sorted from total peritoneal cells. Cells were cultured in the presence of DMOG (1 mM) for 2 h followed by stimulation.

Reaction Conditions

1 mM DMOG for 2 hours

Applications

The hydroxylase inhibitor DMOG reduce susceptibility to endotoxemia by tolerating cells to LPS activation and promoting M2 polarization and subsequent up-regulation of IL-10 by peritoneal B1 cells.

Animal experiment [2]:

Animal models

BALB/c and C57BL/6 mice

Preparation Method

Intraperitoneal injection of DMOG

Dosage form

8 mg DMOG / mouse

Applications

DMOG significantly increased survival after LPS-induced shock. DMOG up-regulated the expression of IL-10 in peritoneal B-1 cells. Mice treated with DMOG before surgery developed severe worsening of disease symptoms and significantly increased mortality.

References:

[1]. Hams E, Saunders SP, et,al. The hydroxylase inhibitor dimethyloxallyl glycine attenuates endotoxic shock via alternative activation of macrophages and IL-10 production by B1 cells. Shock. 2011 Sep;36(3):295-302. doi: 10.1097/SHK.0b013e318225ad7e. PMID: 21844787; PMCID: PMC3157050.

产品描述

DMOG(dihydroxyalanine) is an antagonist of α-ketoglutarate cofactor and an inhibitor of HIF-proline hydroxylase, leading to the stability and accumulation of HIF-1α protein in the nucleus[4].

DMOG reduce susceptibility to endotoxemia by tolerating cells to LPS activation and promoting M2 polarization and subsequent up-regulation of IL-10 by peritoneal B1 cells[1]. DMOG inhibited hydroxyproline synthesis from chick embryo lung, with IC50 values of 9.3 μM and 3.7 μM corresponding to tissue and medium sources, respectively[2].The combination of DMOG and nSi exerted admirable effects on periodontal tissue regeneration. DMOG/nSi-PLGA fibrous membranes could enhance and orchestrate osteogenesis-angiogenesis[4].Inhibition of hydroxylase by oxygen sensing leads to upregulation of the transcription factors HIF-1α and NF-β under normal oxygen in vitro,0.1 to 1mM DMOG can stabilize the expression of HIF-1α[3].DMOG reduces FGF-2-induced proliferation and cyclin A expression by inhibiting prolyl hydroxylase activity in HPASMC[8]. DMOG acts as a pro-angiogenic agent and plays a protective role in experimental model of colitis and diarrhoea via HIF-1 related signal[5][6]. DMOG induces cell autophagy and protect cells from a subsequent OGD insult.[7].

DMOG inhibits endogenous HIF inactivation, and induces angiogenesis in ischaemic skeletal muscles of mice[5]. Up-regulation of hypoxia-inducible factor-1α by DMOG enhances the cardioprotective effects of ischemic postconditioning in hyperlipidemic rats[6].

References:
[1]: Hams E, Saunders SP, et,al. The hydroxylase inhibitor dimethyloxallyl glycine attenuates endotoxic shock via alternative activation of macrophages and IL-10 production by B1 cells. Shock. 2011 Sep;36(3):295-302. doi: 10.1097/SHK.0b013e318225ad7e. PMID: 21844787; PMCID: PMC3157050.
[2]: Baader E, Tschank G, et,al. Inhibition of prolyl 4-hydroxylase by oxalyl amino acid derivatives in vitro, in isolated microsomes and in embryonic chicken tissues. Biochem J. 1994 Jun 1;300 ( Pt 2)(Pt 2):525-30. doi: 10.1042/bj3000525. PMID: 8002959; PMCID: PMC1138193.
[3]: Jaakkola P, Mole DR, et,al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science. 2001 Apr 20;292(5516):468-72. doi: 10.1126/science.1059796. Epub 2001 Apr 5. PMID: 11292861.
[4]: Shang L, Liu Z, Ma B, Shao J, Wang B, Ma C, Ge S. Dimethyloxallyl glycine/nanosilicates-loaded osteogenic/angiogenic difunctional fibrous structure for functional periodontal tissue regeneration. Bioact Mater. 2020 Oct 26;6(4):1175-1188. doi: 10.1016/j.bioactmat.2020.10.010. PMID: 33163699; PMCID: PMC7593348.
[5]: Milkiewicz M, Pugh CW, et,al. Inhibition of endogenous HIF inactivation induces angiogenesis in ischaemic skeletal muscles of mice. J Physiol. 2004 Oct 1;560(Pt 1):21-6. doi: 10.1113/jphysiol.2004.069757. Epub 2004 Aug 19. PMID: 15319416; PMCID: PMC1665195.
[6]: Li X, Zhao H, et,al. Up-regulation of hypoxia-inducible factor-1α enhanced the cardioprotective effects of ischemic postconditioning in hyperlipidemic rats. Acta Biochim Biophys Sin (Shanghai). 2014 Feb;46(2):112-8. doi: 10.1093/abbs/gmt132. Epub 2014 Jan 3. PMID: 24389644.
[7]: Singh A, Wilson JW, et,al. Hypoxia-inducible factor (HIF) prolyl hydroxylase inhibitors induce autophagy and have a protective effect in an in-vitro ischaemia model. Sci Rep. 2020 Jan 31;10(1):1597. doi: 10.1038/s41598-020-58482-w. Erratum in: Sci Rep. 2020 Apr 8;10(1):6041. PMID: 32005890; PMCID: PMC6994562.
[8]: Schultz K, Murthy V, et,al. Prolyl hydroxylase 2 deficiency limits proliferation of vascular smooth muscle cells by hypoxia-inducible factor-1{alpha}-dependent mechanisms. Am J Physiol Lung Cell Mol Physiol. 2009 Jun;296(6):L921-7. doi: 10.1152/ajplung.90393.2008. Epub 2009 Mar 20. PMID: 19304911; PMCID: PMC2692800.

DMOG(二羟基丙氨酸)是 α-酮戊二酸辅助因子的拮抗剂和 HIF-脯氨酸羟化酶的抑制剂,导致 HIF-1α 蛋白在细胞核中的稳定和积累[4]。< /p>\n

DMOG 通过使细胞耐受 LPS 激活并促进 M2 极化和随后腹膜 B1 细胞对 IL-10 的上调来降低对内毒素血症的易感性[1]。 DMOG抑制鸡胚肺中羟脯氨酸的合成,组织来源和培养基来源的IC50值分别为9.3 μM和3.7 μM[2]。DMOG和nSi联合使用对牙周组织再生具有良好的效果. DMOG/nSi-PLGA 纤维膜可增强和协调成骨-血管生成[4]。在体外常氧条件下,氧感应抑制羟化酶导致转录因子 HIF-1α 和 NF-β 上调,0.1 至 1mM DMOG 可以稳定 HIF-1α 的表达[3]。DMOG 通过抑制 HPASMC 中的脯氨酰羟化酶活性来降低 FGF-2 诱导的增殖和细胞周期蛋白 A 的表达[8]< /sup>。 DMOG 作为促血管生成剂,通过 HIF-1 相关信号在结肠炎和腹泻实验模型中发挥保护作用[5][6]。 DMOG 诱导细胞自噬并保护细胞免受随后的 OGD 损伤。[7]

DMOG 抑制内源性 HIF 失活,并在小鼠缺血骨骼肌中诱导血管生成[5]。 DMOG上调缺氧诱导因子1α增强缺血后适应对高脂血症大鼠的心脏保护作用[6]

Chemical Properties

Cas No. 89464-63-1 SDF
别名 二甲基乙二酰氨基乙酸,Dimethyloxallyl Glycine
化学名 methyl 2-[(2-methoxy-2-oxoethyl)amino]-2-oxoacetate
Canonical SMILES COC(=O)CNC(=O)C(=O)OC
分子式 C6H9NO5 分子量 175.14
溶解度 DMF: 30 mg/ml,DMSO: 30 mg/ml,Ethanol: 30 mg/ml,PBS (pH 7.2): 10 mg/ml 储存条件 Store at -20°C
General tips 请根据产品在不同溶剂中的溶解度选择合适的溶剂配制储备液;一旦配成溶液,请分装保存,避免反复冻融造成的产品失效。
储备液的保存方式和期限:-80°C 储存时,请在 6 个月内使用,-20°C 储存时,请在 1 个月内使用。
为了提高溶解度,请将管子加热至37℃,然后在超声波浴中震荡一段时间。
Shipping Condition 评估样品解决方案:配备蓝冰进行发货。所有其他可用尺寸:配备RT,或根据请求配备蓝冰。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 5.7097 mL 28.5486 mL 57.0972 mL
5 mM 1.1419 mL 5.7097 mL 11.4194 mL
10 mM 0.571 mL 2.8549 mL 5.7097 mL
  • 摩尔浓度计算器

  • 稀释计算器

  • 分子量计算器

质量
=
浓度
x
体积
x
分子量
 
 
 
*在配置溶液时,请务必参考产品标签上、MSDS / COA(可在Glpbio的产品页面获得)批次特异的分子量使用本工具。

计算

动物体内配方计算器 (澄清溶液)

第一步:请输入基本实验信息(考虑到实验过程中的损耗,建议多配一只动物的药量)
给药剂量 mg/kg 动物平均体重 g 每只动物给药体积 ul 动物数量
第二步:请输入动物体内配方组成(配方适用于不溶于水的药物;不同批次药物配方比例不同,请联系GLPBIO为您提供正确的澄清溶液配方)
% DMSO % % Tween 80 % saline
计算重置

Research Update

HIF-1α activator DMOG inhibits alveolar bone resorption in murine periodontitis by regulating macrophage polarization

Periodontitis is initiated by serious and sustained bacterial infection and ultimately results in chronic immune-mediated inflammation, tissue destruction, and bone loss. The pathogenesis of periodontitis remains unclear. Host immunological responses to periodontal bacteria ultimately determine the severity and mechanisms governing periodontitis progression. This study aimed to clarify the effect of the hypoxia-inducible factor-1α (HIF-1α) activator dimethyloxalylglycine (DMOG) on a mouse periodontitis model and its underlying role in macrophage polarization. qRT-PCR analysis showed that DMOG inhibited the M1-like polarization of both RAW264.7 macrophages and murine bone marrow macrophages (BMMs) and downregulated TNF-α, IL-6, CD86, and MCP-1 expression in vitro. Immunofluorescence staining and flow cytometry also confirmed the less percentage of F4/80 + CD86 + cells after DMOG treatment. The phosphorylation of NF-κB pathway was also inhibited by DMOG with higher level of HIF-1α expression. Furthermore, mice treated with DMOG showed decreased alveolar bone resorption in the experimental periodontitis model, with significant increases in alveolar bone volume/tissue volume (BV/TV) and bone mineral density (BMD). DMOG treatment of mice decreased the ratio of M1/M2 (CD86+/CD206+) macrophages in periodontal tissues, resulting in the downregulation of proinflammatory cytokines such as TNF-α and IL-6 and increased levels of anti-inflammatory factors such as IL-4 and IL-10. DMOG treatment promoted the number of HIF-1α-positive cells in periodontal tissues. This study demonstrated the cell-specific roles of DMOG in macrophage polarization in vitro and provided insight into the mechanism underlying the protective effect of DMOG in a model of periodontitis.

DMOG Negatively Impacts Tissue Engineered Cartilage Development

Objective: Articular cartilage exists in a hypoxic environment, which motivates the use of hypoxia-simulating chemical agents to improve matrix production in cartilage tissue engineering. The aim of this study was to investigate whether dimethyloxalylglycine (DMOG), a HIF-1α stabilizer, would improve matrix production in 3-dimensional (3D) porcine synovial-derived mesenchymal stem cell (SYN-MSC) co-culture with chondrocytes.
Design: Pellet cultures and scaffold-based engineered cartilage were grown in vitro to determine the impact of chemically simulated hypoxia on 2 types of 3D cell culture. DMOG-treated groups were exposed to DMOG from day 14 to day 21 and grown up to 6 weeks with n = 3 per condition and time point.
Results: The addition of DMOG resulted in HIF-1α stabilization in the exterior of the engineered constructs, which resulted in increased regional type II collagen deposition, but the stabilization did not translate to overall increased extracellular matrix deposition. There was no increase in HIF-1α stabilization in the pellet cultures. DMOG treatment also negatively affected the mechanical competency of the engineered cartilage.
Conclusions: Despite previous studies that demonstrated the efficacy of DMOG, here, short-term treatment with DMOG did not have a uniformly positive impact on the chondrogenic capacity of SYN-MSCs in either pellet culture or in scaffold-based engineered cartilage, as evidenced by reduced matrix production. Such 3D constructs generally have a naturally occurring hypoxic center, which allows for the stabilization of HIF-1α in the interior tissue. Thus, short-term addition of DMOG may not further improve this in cartilage tissue engineered constructs.

Dimethyloxalylglycine (DMOG), a Hypoxia Mimetic Agent, Does Not Replicate a Rat Pheochromocytoma (PC12) Cell Biological Response to Reduced Oxygen Culture

Cells respond to reduced oxygen availability predominately by activation of the hypoxia-inducible factor (HIF) pathway. HIF activation upregulates hundreds of genes that help cells survive in the reduced oxygen environment. The aim of this study is to determine whether chemical-induced HIF accumulation mimics all aspects of the hypoxic response of cells. We compared the effects of dimethyloxalylglycine (DMOG) (a HIF stabiliser) on PC12 cells cultured in air oxygen (20.9% O2, AO) with those cultured in either intermittent 20.9% O2 to 2% O2 (IH) or constant 2% O2 (CN). Cell viability, cell cycle, HIF accumulation, reactive oxygen species (ROS) formation, mitochondrial function and differentiation were used to characterise the PC12 cells and evaluate the impact of DMOG. IH and CN culture reduced the increase in cell numbers after 72 and 96 h and MTT activity after 48 h compared to AO culture. Further, DMOG supplementation in AO induced a dose-dependent reduction in the increase in PC12 cell numbers and MTT activity. IH-cultured PC12 cells displayed increased and sustained HIF-1 expression over 96 h. This was accompanied by increased ROS and mitochondrial burden. PC12 cells in CN displayed little changes in HIF-1 expression or ROS levels. DMOG (0.1 mM) supplementation resulted in an IH-like HIF-1 profile. The mitochondrial burden and action potential of DMOG-supplemented PC12 cells did not mirror those seen in other conditions. DMOG significantly increased S phase cell populations after 72 and 96 h. No significant effect on PC12 cell differentiation was noted with IH and CN culture without induction by nerve growth factor (NGF), while DMOG significantly increased PC12 cell differentiation with and without NGF. In conclusion, DMOG and reduced oxygen levels stabilise HIF and affect mitochondrial activity and cell behaviour. However, DMOG does not provide an accurate replication of the reduced oxygen environments.

Stabilization of HIF-1α alleviates osteoarthritis via enhancing mitophagy

Mitochondrial dysfunction leads to osteoarthritis (OA) and disc degeneration. Hypoxia inducible factor-1α (HIF-1α) mediated mitophagy has a protective role in several diseases. However, the underlying mechanism of HIF-1α mediated mitophagy in OA remains largely unknown. This current study was performed to determine the effect of HIF-1α mediated mitophagy on OA. Therefore, X-ray and tissue staining including HE staining, safranin O-fast green (S-O) and Alcian Blue were used to assess imageology and histomorphology differences of mouse knee joint. Transcriptional analysis was used to find the possible targets in osteoarthritis. Western blot analysis, RT-qPCR and immunofluorescence staining were used to detect the changes in gene and protein levels in the vitro experiment. The expression of HIF-1α was increased in human and mouse OA cartilage. HIF-1α knockdown by siRNA further impair the hypoxia-induced mitochondrial dysfunction; In contrast, HIF-1α mediated protective role was reinforced by prolylhydroxylase (PHD) inhibitor dimethyloxalylglycine (DMOG). In addition, HIF-1α stabilization could alleviate apoptosis and senescence via mitophagy in chondrocytes under hypoxia condition, which could also ameliorate surgery-induced cartilage degradation in mice OA model. In conclusion, HIF-1α mediated mitophagy could alleviate OA, which may serve as a promising strategy for OA treatment.

Dimethyloxalylglycine (DMOG) and the Caspase Inhibitor "Ac-LETD-CHO" Protect Neuronal ND7/23 Cells of Gluocotoxicity

It well known that long-lasting hyperglycaemia disrupts neuronal function and leads to neuropathy and other neurodegenerative diseases. The α-ketoglutarate analogue (DMOG) and the caspase-inhibitor "Ac-LETD-CHO are potential neuroprotective molecules. Whether their protections may also extend glucotoxicity-induced neuropathy is not known. Herein, we evaluated the possible cell-protective effects of DMOG and Ac-LETD-CHO against hyperglycaemia-induced reactive oxygen species and apoptosis in ND7/23 neuronal cells. The impact of glucotoxicity on the expression of HIF-1α and a panel of micro-RNAs of significance in hyperglycaemia and apoptosis was also investigated.ND7/23 cells cultured under hyperglycaemic conditions showed decreased cell viability and elevated levels of ROS production in a dose- and time-dependent manner. However, presence DMOG (500 ?M) and/or Ac-LETD-CHO (50 ?M) counteracted this effect and increase cell viability concomitant with reduction in ROS production, DNA damage and apoptosis. AcLETD-CHO suppressed hyperglycaemia-induced caspase 3 activation in ND7/23 cells. Both DMOG and Ac-LETD-CHO increased HIF-1α expression paralleled with the suppression of miR-126-5p, miR-128-3p and miR-181 expression and upregulation of miR-26b, 106a-5p, 106b-5p, 135a-5p, 135b-5p, 138-5p, 199a-5p, 200a-3p and 200c-3p expression.We demonstrate a mechanistic link for the DMOG and Ac-LETD-CHO protection against hyperglycaemia-induced neuronal dysfunction, DNA damage and apoptosis and thereby propose that pharmacological agents mimicking these effects may represent a promising novel therapy for the hyperglycaemia-induced neuropathy.